Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters










Publication year range
1.
Acta Physiol (Oxf) ; 224(2): e13075, 2018 10.
Article in English | MEDLINE | ID: mdl-29660255

ABSTRACT

AIM: Experimental simulation of near-future ocean acidification (OA) has been demonstrated to affect growth and development of echinoderm larval stages through energy allocation towards ion and pH compensatory processes. To date, it remains largely unknown how major pH regulatory systems and their energetics are affected by trans-generational exposure to near-future acidification levels. METHODS: Here, we used the common sea star Asterias rubens in a reciprocal transplant experiment comprising different combinations of OA scenarios, to study trans-generational plasticity using morphological and physiological endpoints. RESULTS: Acclimation of adults to pHT 7.2 (pCO2 3500 µatm) led to reductions in feeding rates, gonad weight and fecundity. No effects were evident at moderate acidification levels (pHT 7.4; pCO2 2000 µatm). Parental pre-acclimation to pHT 7.2 for 85 days reduced developmental rates even when larvae were raised under moderate and high pH conditions, whereas pre-acclimation to pHT 7.4 did not alter offspring performance. Microelectrode measurements and pharmacological inhibitor studies carried out on larval stages demonstrated that maintenance of alkaline gastric pH represents a substantial energy sink under acidified conditions that may contribute up to 30% to the total energy budget. CONCLUSION: Parental pre-acclimation to acidification levels that are beyond the pH that is encountered by this population in its natural habitat (eg, pHT 7.2) negatively affected larval size and development, potentially through reduced energy transfer. Maintenance of alkaline gastric pH and reductions in maternal energy reserves probably constitute the main factors for a reduced juvenile recruitment of this marine keystone species under simulated OA.


Subject(s)
Acclimatization/physiology , Asterias/physiology , Gastrointestinal Tract/physiology , Homeostasis/physiology , Seawater/chemistry , Animals , Climate Change , Gastrointestinal Tract/chemistry , Humans , Hydrogen-Ion Concentration , Larva
2.
J Exp Biol ; 219(Pt 15): 2300-10, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27489216

ABSTRACT

The excretion of nitrogenous waste products in the form of ammonia (NH3) and ammonium (NH4 (+)) is a fundamental process in aquatic organisms. For mytilid bivalves, little is known about the mechanisms and sites of excretion. This study investigated the localization and the mechanisms of ammonia excretion in mytilid mussels. An Rh protein was found to be abundantly expressed in the apical cell membrane of the plicate organ, which was previously described as a solely respiratory organ. The Rh protein was also expressed in the gill, although at significantly lower concentrations, but was not detectable in mussel kidney. Furthermore, NH3/NH4 (+) was not enriched in the urine, suggesting that kidneys are not involved in active NH3/NH4 (+) excretion. Exposure to elevated seawater pH of 8.5 transiently reduced NH3/NH4 (+) excretion rates, but they returned to control values following 24 h acclimation. These mussels had increased abundance of V-type H(+)-ATPase in the apical membranes of plicate organ cells; however, NH3/NH4 (+) excretion rates were not affected by the V-type H(+)-ATPase specific inhibitor concanamycin A (100 nmol l(-1)). In contrast, inhibition of ciliary beating with dopamine and increased seawater viscosity significantly reduced NH3 excretion rates under control pH (8.0). These results suggest that NH3/NH4 (+) excretion in mytilid mussels takes place by passive NH3 diffusion across respiratory epithelia via the Rh protein, facilitated by the water current produced for filter feeding, which prevents accumulation of NH3 in the boundary layer. This mechanism would be energy efficient for sessile organisms, as they already generate water currents for filter feeding.


Subject(s)
Ammonia/metabolism , Bivalvia/metabolism , Cilia/metabolism , Animal Structures/anatomy & histology , Animal Structures/enzymology , Animals , Bivalvia/enzymology , Epithelium/metabolism , Gills/metabolism , Hemolymph/metabolism , Hydrogen-Ion Concentration , Ion Transport , Proteins/metabolism , Seawater/chemistry , Vacuolar Proton-Translocating ATPases/metabolism
3.
Acta Physiol (Oxf) ; 213(1): 277-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25362991

ABSTRACT

AIMS: Extracellular ATP is an important regulator of renal tubular transport. Recently, we found that basolateral ATP markedly inhibits Na(+) and Cl(-) absorption in mouse medullary thick ascending limb (mTAL) via a P2X receptor. The underlying mechanism that mediates this ATP-dependent transport inhibition in mTAL is, however, unclear. The renal outer medullary K(+) channel (ROMK) is sensitive to intracellular pH where a reduction leads to closing of ROMK. We speculated that P2X receptor stimulation in the TAL could lead to changes in pHi , leading to a reduction in NaCl transport. METHODS: To test this hypothesis, we measured pHi in single perfused mouse mTALs using the fluorescent ratiometric dye 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein acetoxymethylester. RESULTS: Interestingly, basolateral ATP (100 µm) caused a prominent, reversible intracellular alkalization of mTAL, with an average pHi increase of 0.14 ± 0.02 (n = 14). This was completely abolished by the P2X receptor antagonist periodate-oxidized ATP (50 µm). The P2X receptor-mediated intracellular alkalization required the activity of the apical Na(+) /H(+) exchanger (NHE3). Typically, Gq -coupled receptors cause a significant acidification of tubular epithelial cells, which was confirmed in this study, by P2Y2 and Ca(2+) sensing receptor stimulation. CONCLUSION: This study reports that stimulation of basolateral P2X receptors causes a substantial intracellular alkalization in the isolated perfused mouse mTAL. This intracellular alkalization is mediated through an increased apical NHE3 activity, similar to what we previously observed when tubular transport is inhibited with furosemide. This increased NHE3 activity causes H(+) secretion in the mTAL and provides further support that the TAL is a site of urinary acidification.


Subject(s)
Kidney Medulla/metabolism , Loop of Henle/metabolism , Receptors, Purinergic P2X/metabolism , Animals , Cell Separation/methods , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Ion Transport/physiology , Mice , Potassium/metabolism , Sodium/metabolism , Sodium-Hydrogen Exchangers/metabolism
4.
Am J Physiol Renal Physiol ; 302(7): F865-74, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22218592

ABSTRACT

Na(+)-K(+)-2Cl(-) cotransporter (NKCC2)-mediated NaCl reabsorption in the thick ascending limb (TAL) is stimulated by AVP via V2 receptor/PKA/cAMP signaling. This process is antagonized by locally produced eicosanoids such as 20-HETE or prostaglandin E(2), which are synthesized in a phospholipase A(2)-dependent reaction cascade. Using microarray-based gene expression analysis, we found evidence for an AVP-dependent downregulation of the calcium-independent isoform of PLA(2), iPLA(2)ß, in the outer medulla of rats. In the present study, we therefore examined the contribution of iPLA(2)ß to NKCC2 regulation. Immunoreactive iPLA(2)ß protein was detected in cultured mTAL cells as well as in the entire TAL of rodents and humans with the exception of the macula densa. Administration of the V2 receptor-selective agonist desmopressin (5 ng/h; 3 days) to AVP-deficient diabetes insipidus rats increased outer medullary phosphorylated NKCC2 (pNKCC2) levels more than twofold in association with a marked reduction in iPLA(2)ß abundance (-65%; P < 0.05), thus confirming microarray results. Inhibition of iPLA(2)ß in Sprague-Dawley rats with FKGK 11 (0.5 µM) or in mTAL cells with FKGK 11 (10 µM) or (S)-bromoenol lactone (5 µM) for 1 h markedly increased pNKCC2 levels without affecting total NKCC2 expression. Collectively, these data indicate that iPLA(2)ß acts as an inhibitory modulator of NKCC2 activity and suggest that downregulation of iPLA(2)ß may be a relevant step in AVP-mediated urine concentration.


Subject(s)
Group VI Phospholipases A2/metabolism , Loop of Henle/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Vasopressins/metabolism , Animals , Antibodies , Arachidonic Acids , Cells, Cultured , Deamino Arginine Vasopressin , Down-Regulation , Fluorocarbons , Gene Expression , Group VI Phospholipases A2/immunology , Guinea Pigs , Humans , Isoenzymes , Ketones , Kidney Medulla/metabolism , Male , Mice , Mice, Inbred C57BL , Naphthalenes , Organophosphonates , Phosphorylation , Pyrones , Rats , Rats, Brattleboro , Rats, Sprague-Dawley , Solute Carrier Family 12, Member 1
5.
New Phytol ; 190(3): 595-608, 2011 May.
Article in English | MEDLINE | ID: mdl-21294736

ABSTRACT

• To understand the influence of changing surface ocean pH and carbonate chemistry on the coccolithophore Emiliania huxleyi, it is necessary to characterize mechanisms involved in pH homeostasis and ion transport. • Here, we measured effects of changes in seawater carbonate chemistry on the fluorescence emission ratio of BCECF (2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein) as a measure of intracellular pH (pH(i)). Out of equilibrium solutions were used to differentiate between membrane permeation pathways for H(+), CO(2) and HCO(3)(-). • Changes in fluorescence ratio were calibrated in single cells, resulting in a ratio change of 0.78 per pH(i) unit. pH(i) acutely followed the pH of seawater (pH(e)) in a linear fashion between pH(e) values of 6.5 and 9 with a slope of 0.44 per pH(e) unit. pH(i) was nearly insensitive to changes in seawater CO(2) at constant pH(e) and HCO(3)(-). An increase in extracellular HCO(3)(-) resulted in a slight intracellular acidification. In the presence of DIDS (4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid), a broad-spectrum inhibitor of anion exchangers, E. huxleyi acidified irreversibly. DIDS slightly reduced the effect of pH(e) on pH(i). • The data for the first time show the occurrence of a proton permeation pathway in E. huxleyi plasma membrane. pH(i) homeostasis involves a DIDS-sensitive mechanism.


Subject(s)
Cell Membrane Permeability , Cytological Techniques/methods , Haptophyta/cytology , Haptophyta/metabolism , Protons , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Bicarbonates/metabolism , Calibration , Carbon Dioxide/metabolism , Cell Membrane Permeability/drug effects , Fluoresceins/pharmacology , Haptophyta/drug effects , Hydrogen-Ion Concentration/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Kinetics , Nigericin/metabolism , Seawater/chemistry , Solutions
6.
Am J Physiol Renal Physiol ; 298(3): F502-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20007345

ABSTRACT

Vasopressin influences salt and water transport in renal epithelia. This is coordinated by the combined action of V2 receptor-mediated effects along distinct nephron segments. Modulation of NaCl reabsorption by vasopressin has been established in the loop of Henle, but its role in the distal convoluted tubule (DCT), an effective site for fine regulation of urinary electrolyte composition and the target for thiazide diuretics, is largely unknown. The Na+-Cl- cotransporter (NCC) of DCT is activated by luminal trafficking and phosphorylation at conserved NH2-terminal residues. Here, we demonstrate the effects of short-term vasopressin administration (30 min) on NCC activation in Brattleboro rats with central diabetes insipidus (DI) using the V2 receptor agonist desmopressin (dDAVP). The fraction of NCC abundance in the luminal plasma membrane was significantly increased upon dDAVP as shown by confocal microscopy, immunogold cytochemistry, and Western blot, suggesting increased apical trafficking of the transporter. Changes were paralleled by augmented phosphorylation of NCC as detected by antibodies against phospho-threonine and phospho-serine residues (2.5-fold increase at Thr53 and 1.4-fold increase at Ser71). dDAVP-induced phosphorylation of NCC, studied in tubular suspensions in the absence of systemic effects, was enhanced as well (1.7-fold increase at Ser71), which points to the direct mode of action of vasopressin in DCT. Changes were more pronounced in early (DCT1) than in late DCT as distinguished by the distribution of 11beta-hydroxysteroid dehydrogenase 2 in DCT2. These results suggest that the vasopressin-V(2) receptor-NCC signaling cascade is a novel effector system to adjust transepithelial NaCl reabsorption in DCT.


Subject(s)
Antidiuretic Agents/administration & dosage , Cell Membrane/drug effects , Deamino Arginine Vasopressin/administration & dosage , Diabetes Insipidus, Neurogenic/metabolism , Kidney Tubules, Distal/drug effects , Receptors, Drug/drug effects , Receptors, Vasopressin/agonists , Sodium Chloride Symporter Inhibitors/pharmacology , Symporters/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Chlorides/metabolism , Diabetes Insipidus, Neurogenic/drug therapy , Diabetes Insipidus, Neurogenic/pathology , Diabetes Insipidus, Neurogenic/physiopathology , Disease Models, Animal , Immunohistochemistry , Kidney Tubules, Distal/metabolism , Kidney Tubules, Distal/physiopathology , Kidney Tubules, Distal/ultrastructure , Male , Microscopy, Confocal , Natriuresis/drug effects , Phosphorylation , Protein Transport , Rats , Rats, Brattleboro , Rats, Wistar , Receptors, Drug/metabolism , Receptors, Vasopressin/metabolism , Sodium/metabolism , Solute Carrier Family 12, Member 3 , Symporters/metabolism , Time Factors , Up-Regulation
7.
Diabetologia ; 48(8): 1645-53, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16010524

ABSTRACT

AIMS/HYPOTHESIS: Renal accumulation of AGEs may contribute to the progression of diabetic nephropathy. We evaluated the effect of ramipril (a pure ACE inhibitor) and AVE7688 (a dual inhibitor of ACE and neutral endopeptidase) on renal accumulation of the advanced glycation end-product (AGE) 3-deoxyglucosone-imidazolone, carboxymethyllysine (CML) and pentosidine, and on clearance of CML in type 2 diabetes. METHODS: Male Zucker diabetic fatty rats (ZDF, Gmi-fa/fa) rats were treated from age 10 to 37 weeks with ramipril (1 mg.kg(-1).day(-1)), AVE7688 (45 mg.kg(-1).day(-1)) or without drug. Ramipril and AVE7688 reduced albuminuria by 30 and 90%, respectively. RESULTS: ZDF rats showed increased renal accumulation of the AGE subtypes 3-deoxyglucosone-imidazolone, pentosidine and CML by about 40, 55 and 55%, respectively compared with heterozygous, non-diabetic control animals at the age of 37 weeks. AVE7688 but not ramipril attenuated the renal accumulation of 3-deoxyglucosone-imidazolone, pentosidine and CML and improved CML clearance in ZDF rats. During glycation reactions in vitro, AVE7688 also demonstrated potent chelating activity and inhibited metal-catalysed formation of pentosidine and CML. CONCLUSIONS/INTERPRETATION: Improved AGE clearance and direct inhibition of AGE formation by chelation may contribute to reduced accumulation of renal AGEs and to the nephroprotective effects of vasopeptidase inhibition in type 2 diabetes.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Glycation End Products, Advanced/metabolism , Kidney/metabolism , Protease Inhibitors/pharmacology , Animals , Ascorbic Acid/metabolism , Blood Glucose/metabolism , Chromatography, High Pressure Liquid , Creatine/metabolism , Deoxyglucose/analogs & derivatives , Deoxyglucose/metabolism , Glycated Hemoglobin/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , Kidney/drug effects , Lysine/analogs & derivatives , Lysine/metabolism , Male , Oxidation-Reduction , Ramipril/pharmacology , Rats , Rats, Zucker , Spectrometry, Fluorescence
8.
Naunyn Schmiedebergs Arch Pharmacol ; 370(3): 183-92, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15340774

ABSTRACT

Previous studies in pigs and goats have demonstrated that AVE0118 prolongs atrial refractoriness without any effect on the QT-interval. The purpose of the present study was to investigate the effect of the compound on various cardiac ion channels. AVE0118 blocked the pig Kv1.5 and the human Kv1.5 expressed in Xenopus oocytes with IC(50) values of 5.4+/-0.7 microM and 6.2+/-0.4 microM respectively. In Chinese hamster ovary (CHO) cells, AVE0118 decreased the steady-state hKv1.5 current with an IC(50) of 1.1+/-0.2 microM. The hKv4.3/KChIP2.2 current in CHO cells was blocked by AVE0118 by accelerating the apparent time-constant of inactivation ( tau(inact)), and the integral current was inhibited with an IC(50) of 3.4+/-0.5 microM. At 10 microM AVE0118 tau(inact) decreased from 9.3+/-0.6 ms ( n=8, control) to 3.0+/-0.3 ms ( n=8). The K(ACh) current was investigated in isolated pig atrial myocytes by application of 10 microM carbachol. At a clamp potential of -100 mV the I(KACh) was half-maximally blocked by 4.5+/-1.6 microM AVE0118. In the absence of carbachol, AVE0118 had no effect on the inward current recorded at -100 mV. Effects on the I(Kr) current were investigated on HERG channels expressed in CHO cells. AVE0118 blocked this current half-maximally at approximately 10 microM. Comparable results were obtained in isolated guinea pig ventricular myocytes, where half-maximal inhibition of the I(Kr) tail current occurred at a similar concentration of AVE0118. Other ionic currents, like the I(Ks), I(KATP) (recorded in guinea pig ventricular myocytes), and L-type Ca(2+) (recorded in pig atrial myocytes) were blocked by 10 microM AVE0118 by 10+/-3% ( n=6), 28+/-7% ( n=4), and 22+/-13% ( n=5) respectively. In summary, AVE0118 preferentially inhibits the atrial K(+) channels I(Kur), I(to) and I(KACH). This profile may explain the selective prolongation of atrial refractoriness described previously in pigs and goats.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Carbachol/pharmacology , Cardiotonic Agents/pharmacology , Ion Channels/drug effects , Myocytes, Cardiac/drug effects , Animals , CHO Cells , Calcium-Binding Proteins/antagonists & inhibitors , Cells, Cultured , Cricetinae , Cricetulus , Electrophysiology , Humans , Kv Channel-Interacting Proteins , Kv1.5 Potassium Channel , Molecular Biology , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Shal Potassium Channels , Swine , Xenopus
9.
Naunyn Schmiedebergs Arch Pharmacol ; 369(4): 374-81, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15024553

ABSTRACT

During heart ischemia, ATP-sensitive potassium channels in the sarcolemmal membrane (sarcK(ATP)) open and cause shortening of the action potential duration. This creates heterogeneity of repolarization, being responsible for the development of re-entry arrhythmias and sudden cardiac death. Therefore, the aim is to develop selective blockers of the cardiac sarcK(ATP) channel. In the present study we established an in vitro model and classified 5 K(ATP) channel inhibitors with respect to their potency and selectivity between cardiomyocytes and the coronary vasculature and compared the results with inhibition of Kir6.2/SUR2A channels expressed in HEK293 cells, recorded with the Rb(+)-efflux methods. We used Langendorff-perfused guinea pig hearts, where low-flow ischemia plus hypoxia was performed by reducing the coronary flow (CF) to 1.2 ml/min and by gassing the perfusion solution with N(2) instead of O(2). Throughout the experiment, the monophasic action potential duration at 90% repolarization (MAPD(90)) was recorded. In separate experiments, high-flow hypoxia was produced by oxygen reduction in the perfusate from 95% to 20%, which caused an increase in the coronary flow. Under normoxic conditions, the substances glibenclamide, repaglinide, meglitinide, HMR 1402 and HMR 1098 (1 microM each) reduced the CF by 34%, 38%, 19%, 12% and 5%, respectively. The hypoxia-induced increase in CF was inhibited by the compounds half-maximally at 25 nM, approximately 200 nM, 600 nM, approximately 9 microM and >100 microM, respectively. In control experiments after 5 min low-flow ischemia plus hypoxia, the MAPD(90) shortened from 121+/-2 to 99+/-2 ms ( n=29). This shortening was half-maximally inhibited by the substances at concentrations of 95 nM, 74 nM, 400 nM, 110 nM and 550 nM, respectively. In HEK293 cells the Rb(+)-efflux through KIR6.2/SUR2A channels was inhibited by the compounds with IC(50) values of 21 nM, 67 nM, 205 nM, 60 nM and 181 nM, respectively. In summary, the present data demonstrate that the sulfonylurea glibenclamide, and the carbamoylbenzoic acid derivatives repaglinide and meglitinide are unselective blockers of K(ATP) channels in cardiac cells and in the cardiac vascular system, whereas the sulfonylthioureas HMR 1402, and especially HMR 1098 selectively blocked the cardiac sarcK(ATP) channel. Blockade of Kir6.2/SUR2A channels in HEK293 cells occurred with comparable efficacy as in the cardiac tissue, indicating that the expression system is suited for screening for novel inhibitors.


Subject(s)
Adenosine Triphosphate/physiology , Heart/drug effects , Potassium Channel Blockers/adverse effects , Potassium Channels/physiology , ATP-Binding Cassette Transporters/physiology , Action Potentials/drug effects , Animals , Cell Line , Coronary Circulation/drug effects , Dose-Response Relationship, Drug , Guinea Pigs , Heart/physiology , Humans , Hypoxia/complications , Hypoxia/physiopathology , In Vitro Techniques , Male , Myocardial Ischemia/complications , Myocardial Ischemia/physiopathology , Potassium Channel Blockers/administration & dosage , Potassium Channels, Inwardly Rectifying/physiology , Receptors, Drug/physiology , Sulfonylurea Receptors
10.
Diabetologia ; 47(1): 98-103, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14618238

ABSTRACT

AIM/HYPOTHESIS: Pharmacological inhibition of the renin angiotensin system has proven clinical efficacy in nephropathies of various origins, including diabetic nephropathy. We tested the effects of the dual inhibition of both angiotensin converting enzyme and neutral endopeptidase by the vasopeptidase inhibitor AVE7688 in an animal model of Type 2 diabetic nephropathy. METHODS: We treated 56 obese Zucker diabetic fatty (ZDF, Gmi-fa/fa) rats aged 34-weeks with either placebo ( n=9) or the vasopeptidase inhibitor AVE7688 in four different doses (each n=9; 3, 10, 30, or 60 mg/kg/d in chow). We used 11 heterozygous (+/fa) rats which received placebo and served as non-diabetic, lean controls. Urinary albumin/creatinine ratio was assessed as a marker of nephropathy at baseline (age 34-weeks) and after 10 weeks of chronic treatment. RESULTS: All obese animals had established diabetes mellitus that was not influenced by AVE7688 (HbA(1c) >12%, stable in all dose groups). There was massive albuminuria in the homozygous ZDF rats (albumine/creatinine ratio >20 mg/mg vs minimal albuminuria in lean controls) that was decreased by AVE7688 in a dose dependent manner (Placebo 2.0+/-4.4 vs 11.9+/-1.8, 13.4+/-0.7, 13.6+/-2.8, and 19.8+/-2.8 mg/mg in the 3, 10, 30, and 60 mg/kg/d groups, respectively; all treatment groups p<0.05 vs Placebo). CONCLUSION/INTERPRETATION: AVE7688 ameliorates proteinuria in Zucker diabetic fatty rats with established diabetes mellitus. Vasopeptidase inhibition represents an effective novel therapeutic principle for intervention in Type 2 diabetic nephropathy independent of metabolic control.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/drug therapy , Heterocyclic Compounds, 3-Ring/therapeutic use , Albuminuria/prevention & control , Animals , Body Weight , Diabetes Mellitus/drug therapy , Drinking Behavior , Energy Intake , Glycated Hemoglobin/metabolism , Male , Obesity , Prodrugs/therapeutic use , Protease Inhibitors/therapeutic use , Rats , Rats, Zucker
11.
J Membr Biol ; 186(2): 89-100, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11944086

ABSTRACT

Thimerosal (o-Ethylmercurithio)benzoic acid, TMS), a membrane-impermeable, sulfhydryl-oxidizing agent, has been described to increase the K+ current IKs in KCNE1-injected Xenopus laevis oocytes. Since there are no cysteine residues in the extracellular domain of KCNE1, it has been proposed that TMS interacts with its partner protein KCNQ1. The aim of this study was therefore to investigate the interaction of TMS with KCNQ1 and the respective K+current IK. In CHO cells stably transfected with KCNQ1/KCNE1, TMS increased IKs, whereas in CHO cells expressing KCNQ1 alone, TMS initially decreased IK. TMS also affected the cytosolic pH (pHi) and the cytosolic Ca2+ activity ([Ca2+]i) in these cells. TMS slowly decreased pHi. With a short delay, TMS increased [Ca2+]i by store depletion and capacitative influx. The time course of the effects of TMS on pHi and [Ca2+]i did not correlate with the effect of TMS on IK. We therefore anticipated a different mode of action by TMS and investigated the influence of TMS on cysteine residues of KCNQ1. For this purpose, KCNQ1wt and two mutants lacking a cysteine residue in the S6 or the S3 segment (KCNQ1C331A and KCNQ1C214A, respectively) were expressed in Xenopus laevis oocytes. A sustained current decrease was observed in KCNQ1wt and KCNQ1C331A, but not in KCNQ1C214A-injected oocytes. The analysis of tail currents, I/V curves and activation kinetics revealed a complex effect of TMS on the gating of KCNQ1wt and KCNQ1C331A. In another series we investigated the effect of TMS on IKs. TMS increased IKs of KCNQ1C214A/KCNE1-injected oocytes significantly less than IKs in KCNQ1wt/KCNE1- or KCNQ1C331A/KCNE1-injected cells. These results suggest that thimerosal interacts with the cysteine residue C214 in the S3 segment of KCNQ1, leading to a change of its gating properties. Our results support the idea that not only the inner shell, but also the outer shell of the channel is important for the gating behavior of voltage dependent K+ channels.


Subject(s)
Ion Channel Gating/drug effects , Oxidants/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Thimerosal/pharmacology , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Electric Conductivity , Humans , Hydrogen-Ion Concentration , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Oocytes , Potassium Channels/genetics , Xenopus laevis
12.
Pflugers Arch ; 443(5-6): 822-8, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11889581

ABSTRACT

KCNE1 (IsK, minK) co-assembles with KCNQ1 (KvLQT1) to form voltage-dependent K(+) channels. Both KCNQ1 and KCNE1 are expressed in epithelial cells of gut and exocrine pancreas. We examined the role of KCNQ1/KCNE1 in Cl(-) secretion in small and large intestine and exocrine pancreas using the KCNE1 knockout mouse. Immunofluorescence revealed a similar basolateral localization of KCNQ1 in jejunum and colon of KCNE1 wild-type and knockout mice. Electrogenic Cl(-) secretion in the colon was not affected by gene disruption of KCNE1; in jejunum forskolin-induced short-circuit current was some 40% smaller but without being significantly different. Inhibition of KCNQ1 channels by 293B (IC(50) 1 micromol l(-1)) and by IKS224 (IC(50) 14 nmol l(-1)) strongly diminished intestinal Cl(-) secretion. In exocrine pancreas of wild-type mice, KCNQ1 was predominantly located at the basolateral membrane. In KCNE1 knockout mice, however, the basolateral staining was less pronounced and the distribution of secretory granules was irregular. A slowly activating and 293B-sensitive K(+) current was activated via cholinergic stimulation in pancreatic acinar cells of wild-type mice. In KCNE1 knockout mice this K(+) current was strongly reduced. In conclusion intestinal Cl(-) secretion is independent from KCNE1 but requires KCNQ1. In mouse pancreatic acini KCNQ1 probably co-assembled with KCNE1 leads to a voltage-dependent K(+) current that might be of importance for electrolyte and enzyme secretion.


Subject(s)
Intestinal Mucosa/metabolism , Pancreas/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Potassium Channels/metabolism , Animals , Chlorides/metabolism , Colon/metabolism , Intestinal Mucosa/chemistry , Jejunum/metabolism , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Membrane Potentials/physiology , Mice , Mice, Knockout , Pancreas/chemistry , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels/analysis
13.
Pflugers Arch ; 443(2): 297-305, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11713657

ABSTRACT

Epithelial Na+ channel (ENaC) activity in kidney and colon is stimulated by aldosterone acting on the mineralocorticoid receptor (MR). MR and the glucocorticoid receptor (GR) show high homology in their DNA-binding domain and have similar affinities to mineralo- and glucocorticoids. We therefore asked whether the glucocorticoid-mediated activation of ENaC is restricted to the presence of MR and used the MR knockout mouse model to address this question. Due to their MR deficiency and the consecutive reduction of ENaC activity these mice die as neonates, and even after appropriate substitution therapy adult MR knockout mice suffer from high Na+ loss and hyperkalemia. In the present study, glucocorticoid treatment restored plasma K+ and almost normalized the fractional excretions of Na+ (FENa+) and K+ (FEK+) in adult salt-substituted MR knockout mice, while the effect of amiloride on FENa+ and FEK+ was augmented in these animals. In order to estimate ENaC activity, measurements of transepithelial equivalent short-circuit current (Isc) were performed. Glucocorticoids induced an amiloride-sensitive Na+ absorption in renal cortical collecting duct and distal colon of MR-/- of about 25% and 50% of the currents observed in glucocorticoid-treated wild-type mice, respectively. In the colon glucocorticoid treatment increased the mRNA abundance of all three ENaC subunits, in the kidney only alpha-ENaC was increased. The regulation of ENaC expression was the same in both genotypes and thus irrespective of the presence of MR. These data show that MR is no prerequisite for the activation of ENaC transcription and activity, and that the respective mechanisms can be stimulated via GR.


Subject(s)
Glucocorticoids/pharmacology , Receptors, Mineralocorticoid/physiology , Sodium Channels/metabolism , Triamcinolone/pharmacology , Amiloride/pharmacology , Animals , Animals, Newborn/urine , Blood Pressure/drug effects , Body Water/metabolism , Colon/drug effects , Colon/metabolism , Corticosterone/urine , Diuretics/pharmacology , Electrolytes/blood , Epithelial Sodium Channels , Homeostasis/drug effects , In Vitro Techniques , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/metabolism , Mice , Mice, Knockout/genetics , RNA, Messenger/metabolism , Receptors, Mineralocorticoid/deficiency , Receptors, Mineralocorticoid/genetics , Sodium Channels/genetics
14.
Pflugers Arch ; 443(1): 146-54, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11692278

ABSTRACT

KCNQ1 (KVLQT1) K+ channels play an important role during electrolyte secretion in airways and colon. KCNQ1 was cloned recently from NaCl-secreting shark rectal glands. Here we study the properties and regulation of the cloned sKVLQT1 expressed in Xenopus oocytes and Chinese hamster ovary (CHO) cells and compare the results with those obtained from in vitro perfused rectal gland tubules (RGT). The expression of sKCNQ1 induced voltage-dependent, delayed activated K+ currents, which were augmented by an increase in intracellular cAMP and Ca2+. The chromanol derivatives 293B and 526B potently inhibited sKCNQ1 expressed in oocytes and CHO cells, but had little effect on RGT electrolyte transport. Short-circuit currents in RGT were activated by alkalinization and were decreased by acidification. In CHO cells an alkaline pH activated and an acidic pH inhibited 293B-sensitive KCNQ1 currents. Noise analysis of the cell-attached basolateral membrane of RGT indicated the presence of low-conductance (<3 pS) K+ channels, in parallel with other K+ channels. sKCNQ1 generated similar small-conductance K+ channels upon expression in CHO cells and Xenopus oocytes. The results suggest the presence of low-conductance KCNQ1 K+ channels in RGT, which are probably regulated by changes in intracellular cAMP, Ca2+ and pH.


Subject(s)
Dogfish , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Salt Gland/chemistry , Animals , CHO Cells , Calcium/pharmacology , Cricetinae , Cyclic AMP/pharmacology , Electric Conductivity , Female , Gene Expression , Hydrogen-Ion Concentration , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels/genetics , Transfection , Xenopus laevis
15.
J Med Chem ; 44(23): 3831-7, 2001 Nov 08.
Article in English | MEDLINE | ID: mdl-11689069

ABSTRACT

Since the discovery of the I(Ks)-potassium channel as the slowly activating component of the delayed rectifier current (I(k)) in cardiac tissue, the search for blockers of this current has been intense. During the screening of K(ATP)-channel openers of the chromanol type we found that chromanol 293B was able to block I(Ks). Chromanol 293B is a sulfonamide analogue of the K(ATP)-channel openers but had no activity on this target. Experiments were initiated to improve the activity and properties based on this lead compound. As a screening model we used Xenopus oocytes injected with human minK (KCNE1). Variations of the aromatic substituent and the sulfonamide group were prepared, and their activity was evaluated. We found that the greatest influence on activity was found in the aromatic substituents. The most active compounds were alkoxy substituted. We chose HMR1556 ((3R, 4S)-(+)-N-[-3-hydroxy-2,2-dimethyl-6-(4,4,4-trifluorobutoxy)chroman-4-yl]-N-methyl-ethanesulfonamide) 10a for development as an antiarrhythmic drug. The absolute configuration, resulting from an X-ray single-crystal structure analysis, was determined.


Subject(s)
Chromans/chemical synthesis , Potassium Channel Blockers , Potassium Channel Blockers/chemical synthesis , Potassium Channels, Voltage-Gated , Sulfonamides/chemical synthesis , Animals , Chromans/chemistry , Chromans/pharmacology , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , In Vitro Techniques , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Xenopus laevis
16.
J Physiol ; 535(Pt 2): 349-58, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11533128

ABSTRACT

1. The secretagogue-activated K(+) conductance is indispensable for the electrogenic Cl(-) secretion in exocrine tissue. In this study, we investigated the effect of secretin and other cAMP-mediated secretagogues on the slowly activating voltage-dependent K(+) current (I(Ks)) of rat pancreatic acinar cells (RPAs) with the whole-cell patch clamp technique. 2. Upon depolarization, RPAs showed I(Ks) superimposed upon the instantaneous background outward current. Secretin (5 nM), vasoactive intestinal peptide (5 nM), forskolin (5 microM), isoprenaline (10 microM) or 3-isobutyl-1-methylxanthine (IBMX, 0.1 mM) increased the amplitude of I(Ks) two- to fourfold. 3. The physiological concentration of secretin (50 pM) had a relatively weak effect on I(Ks) (160 % increase), which was significantly enhanced by transient co-stimulation with carbachol (CCh) (10 microM). However, the secretin-induced production of cAMP, which was measured by enzyme-linked immunosorbent assay, was not augmented by co-stimulation with CCh. 4. This study is the first to demonstrate the regulation of K(+) channels in RPAs by cAMP-mediated agonists. The I(Ks) channel is a common target for both Ca(2+) and cAMP agonists. The vagal stimulation under the physiological concentration of secretin facilitates I(Ks), which provides an additional driving force for Cl(-) secretion.


Subject(s)
Pancreas/metabolism , Potassium Channels/metabolism , Secretin/pharmacology , Sulfonamides , 1-Methyl-3-isobutylxanthine/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Calcium Signaling/physiology , Carbachol/pharmacology , Chlorides/metabolism , Cholinergic Agonists/pharmacology , Colforsin/pharmacology , Cyclic AMP/metabolism , Enzyme Inhibitors/pharmacology , Gastrointestinal Agents/pharmacology , Isoproterenol/pharmacology , Isoquinolines/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Pancreas/cytology , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Rats , Secretin/metabolism , Vasoactive Intestinal Peptide/pharmacology
17.
J Biol Chem ; 276(45): 42268-75, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11527966

ABSTRACT

The gene KCNQ1 encodes a K(+) channel alpha-subunit important for cardiac repolarization, formerly known as K(v)LQT1. In large and small intestine a channel complex consisting of KCNQ1 and the beta-subunit KCNE3 (MiRP2) is known to mediate the cAMP-activated basolateral K(+) current, which is essential for luminal Cl(-) secretion. Northern blot experiments revealed an expression of both subunits in lung tissue. However, previous reports suggested a role of KCNE1 (minK, Isk) but not KCNE3 in airway epithelial cells. Here we give evidence that KCNE1 is not detected in murine tracheal epithelial cells and that Cl(-) secretion by these cells is not reduced by the knock-out of the KCNE1 gene. In contrast we show that a complex consisting of KCNQ1 and KCNE3 probably forms a basolateral K(+) channel in murine tracheal epithelial cells. As described for colonic epithelium, the current through KCNQ1 complexes in murine trachea is specifically inhibited by the chromanol 293B. A 293B-sensitive current was present after stimulation with forskolin and agonists that increase Ca(2+) as well as after administration of the pharmacological K(+) channel activator, 1-EBIO. A 293B-inhibitable current was already present under control conditions and reduced after administration of amiloride indicating a role of this K(+) channel not only for Cl(-) secretion but also for Na(+) reabsorption. We conclude that at least in mice a KCNQ1 channel complex seems to be the dominant basolateral K(+) conductance in tracheal epithelial cells.


Subject(s)
Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Trachea/chemistry , Adenosine Triphosphate/pharmacology , Animals , Calcium/metabolism , Chlorides/metabolism , Cyclic AMP/physiology , Indoles/pharmacology , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium Channels/chemistry , Potassium Channels/genetics , Protein Subunits
18.
Cell Physiol Biochem ; 11(4): 219-30, 2001.
Article in English | MEDLINE | ID: mdl-11509830

ABSTRACT

BACKGROUND AND AIMS: The Ca(2+)-activated K(+) channel rSK4 is the rat homologue of the human SK4/IK1 (KCNN4) channel. In colonic mucosa rSK4 plays a key role during acetylcholin-induced secretion. This study was aimed to characterize the properties of the rat SK4 channel. METHODS: Electrophysiological measurements were performed on rSK4 expressing Xenopus laevis oocytes and rat colonic crypts. Intracellular Ca(2+) activity was assessed by Oregon Green fluorescence measurements. RESULTS: The 10 pS rSK4 expressed in oocytes was Ca(2+)-sensitive and inhibited by calmodulin antagonists. 1-ethyl-2-benzimidazolinone (1-EBIO), a known activator of SK4/IK1 channels, also activated rSK4. 1-EBIO affected the current neither at saturating Ca(2+) activities nor under Ca(2+)-free conditions, but increased the Ca(2+) sensitivity of rSK4. rSK4 was strongly activated by cytosolic ATP. However, PKA itself, PKA inhibitors and mutation of the PKA phosphorylation site (S332A) did not affect channel activity. The PKC activator 1,2-dioctanoyl-sn-glycerol and the PKC inhibitor bisindolylmaleimide also failed to influence rSK4. CONCLUSION: The Ca(2+)-sensitive rSK4 is activated by 1-EBIO probably via facilitation of the Ca(2+)-calmodulin-rSK4 interaction. The strong ATP-activation of rSK4 is likely to be caused by phosphorylation via a yet unknown kinase and might involve additional subunits.


Subject(s)
Intestinal Mucosa/metabolism , Potassium Channels, Calcium-Activated , Potassium Channels/metabolism , Potassium/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Benzimidazoles/pharmacology , Calcium/metabolism , Calcium/pharmacology , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Charybdotoxin/pharmacology , Colforsin/pharmacology , Colon/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Intermediate-Conductance Calcium-Activated Potassium Channels , Ionomycin/pharmacology , Oocytes/metabolism , Patch-Clamp Techniques , Phosphorylation , Potassium Channels/drug effects , Protein Kinase C/metabolism , Rats , Recombinant Proteins/metabolism , Sulfonamides/pharmacology , Xenopus laevis
19.
Proc Natl Acad Sci U S A ; 98(15): 8792-7, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11438691

ABSTRACT

The voltage-dependent K(+) channel responsible for the slowly activating delayed K(+) current I(Ks) is composed of pore-forming KCNQ1 and regulatory KCNE1 subunits, which are mutated in familial forms of cardiac long QT syndrome. Because KCNQ1 and KCNE1 genes also are expressed in epithelial tissues, such as the kidneys and the intestine, we have investigated the adaptation of KCNE1-deficient mice to different K(+) and Na(+) intakes. On a normal K(+) diet, homozygous kcne1(-/-) mice exhibit signs of chronic volume depletion associated with fecal Na(+) and K(+) wasting and have lower plasma K(+) concentration and higher levels of aldosterone than wild-type mice. Although plasma aldosterone can be suppressed by low K(+) diets or stimulated by low Na(+) diets, a high K(+) diet provokes a tremendous increase of plasma aldosterone levels in kcne1(-/-) mice as compared with wild-type mice (7.1-fold vs. 1.8-fold) despite lower plasma K(+) in kcne1(-/-) mice. This exacerbated aldosterone production in kcne1(-/-) mice is accompanied by an abnormally high plasma renin concentration, which could partly explain the hyperaldosteronism. In addition, we found that KCNE1 and KCNQ1 mRNAs are expressed in the zona glomerulosa of adrenal glands where I(Ks) may directly participate in the control of aldosterone production by plasma K(+). These results, which show that KCNE1 and I(Ks) are involved in K(+) homeostasis, might have important implications for patients with I(Ks)-related long QT syndrome, because hypokalemia is a well known risk factor for the occurrence of torsades de pointes ventricular arrhythmia.


Subject(s)
Aldosterone/metabolism , Long QT Syndrome/congenital , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Potassium/metabolism , Aldosterone/blood , Animals , Blood Pressure , Colon/metabolism , Disease Models, Animal , Electrocardiography , Feces , Gene Expression , Humans , Ions/metabolism , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Long QT Syndrome/metabolism , Mice , Mice, Knockout , Potassium/blood , Potassium Channels/genetics , Renin/blood , Sodium/metabolism , Sodium/urine , Tissue Distribution
20.
Gastroenterology ; 120(6): 1363-71, 2001 May.
Article in English | MEDLINE | ID: mdl-11313306

ABSTRACT

BACKGROUND & AIMS: Gastric H+ secretion via the H+/K+-adenosine triphosphatase is coupled to the uptake of K+. However, the molecular identity of luminal K+ channels enabling K+ recycling in parietal cells is unknown. This study was aimed to investigate these luminal K+ channels. METHODS: Acid secretion was measured in vivo and in vitro; KCNQ1 protein localization was assessed by immunofluorescence, and acid-sensitivity of KCNQ1 by patch-clamp. RESULTS: We identified KCNQ1, which is mutated in cardiac long QT syndrome, as a K+ channel located in tubulovesicles and apical membrane of parietal cells, where it colocalized with H+/K+-adenosine triphosphatase. Blockade of KCNQ1 current by 293B led to complete inhibition of acid secretion. The putative KCNQ1 subunits, KCNE2 and KCNE3, were abundant in human stomach; KCNE1, however, was absent. Coexpression of KCNE3/KCNQ1 in COS cells led to an acid-insensitive current; KCNE2/KCNQ1 was activated by low extracellular pH. CONCLUSIONS: We identified KCNQ1 as the missing luminal K+ channel in parietal cells and characterized its crucial role in acid secretion. Because KCNE3 and KCNE2 are expressed in human stomach, one or both are candidates to coassemble with KCNQ1 in parietal cells. Thus, stomach- and subunit-specific inhibitors of KCNQ1 might offer new therapeutical perspectives for peptic ulcer disease.


Subject(s)
Gastric Acid/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Dogs , Gastric Acidity Determination , Histamine/pharmacology , Humans , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Male , Mice , Molecular Sequence Data , Potassium Channel Blockers , Potassium Channels/analysis , Rabbits , Rats , Rats, Sprague-Dawley , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...